EP1555035A2 - Matrice de collagène pour l'augmentation de tissus mous - Google Patents

Matrice de collagène pour l'augmentation de tissus mous Download PDF

Info

Publication number
EP1555035A2
EP1555035A2 EP04258168A EP04258168A EP1555035A2 EP 1555035 A2 EP1555035 A2 EP 1555035A2 EP 04258168 A EP04258168 A EP 04258168A EP 04258168 A EP04258168 A EP 04258168A EP 1555035 A2 EP1555035 A2 EP 1555035A2
Authority
EP
European Patent Office
Prior art keywords
implant
collagen
tissue
sulfate
glycosaminoglycan
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP04258168A
Other languages
German (de)
English (en)
Inventor
Lynetta Freeman
Susan Roweton
Ben Walthall
Kien Nguyen
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ethicon Endo Surgery Inc
Original Assignee
Ethicon Endo Surgery Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ethicon Endo Surgery Inc filed Critical Ethicon Endo Surgery Inc
Publication of EP1555035A2 publication Critical patent/EP1555035A2/fr
Withdrawn legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/50Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L27/58Materials at least partially resorbable by the body
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/40Composite materials, i.e. containing one material dispersed in a matrix of the same or different material
    • A61L27/44Composite materials, i.e. containing one material dispersed in a matrix of the same or different material having a macromolecular matrix
    • A61L27/48Composite materials, i.e. containing one material dispersed in a matrix of the same or different material having a macromolecular matrix with macromolecular fillers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/50Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L27/56Porous materials, e.g. foams or sponges

Definitions

  • the present invention relates to reformation of soft tissues within the body. More particularly, the invention relates to compositions useful in reforming the shape of soft tissues and methods of using such compositions in reforming soft tissues.
  • Silicone has been used but can displace and harden over time.
  • Plastic and metal implants have also been used. However, implants such as these may not have a "natural" look or feel, especially as the body changes over time.
  • the invention features devices and methods for soft tissue substitute.
  • the present invention features materials that may be implanted into soft body tissue for correction of soft tissue defects or for soft tissue augmentation.
  • the material comprises a biologically-compatible lattice or matrix.
  • the material may be combined with collagen, proteins or other matrix materials.
  • the matrix is absorbable, biocompatible, used as an acellular or cellular substrate, supports native tissue in-growth in three dimensions, and maintains the biomechanical properties of tissue (e.g., breast tissue) during the postoperative period.
  • tissue e.g., breast tissue
  • the highly porous nature of this product serves as a support structure into which vessels and mesenchymal cells from the wound site migrate. The cells and vessels create new tissue that replaces the collagen glycosaminoglycan as it biodegrades. Because cells invade the matrix material, fibrous encapsulation does not occur to any great degree.
  • cells may be seeded on the lattice substrate.
  • Such cells may be adipocytes (fat cells) and/or preadipocytes (fat cell precursors that differentiate into adipocytes) derived from fat storage areas in the body or fat cells derived from bone marrow, stem cells, or mesenchymal cells that are harvested and developed under laboratory conditions and seeded onto the matrix prior to implantation.
  • the collagen glycosaminoglycan material is placed directly into the wound following surgery to fill the cavity and correct contour defects resulting from the removal of tissue.
  • the product may be used at the time of the initial surgery, or in subsequent surgery.
  • the material used in this invention is preferably in sheet form.
  • the sheet may be randomly folded, rolled, cinched, or configured otherwise to fill the defect.
  • Other forms of the collagen glycosaminoglycan include forms such as blocks (e.g., 2.5 cm x 2.5 cm x 1 cm blocks), spheres, and other configurations.
  • Another object of the invention is to minimize patient discomfort, risk of infection and side effects of repeated medical procedures.
  • the implant material may incorporate radio-opaque materials.
  • the present invention is a soft tissue implant material comprising biologically-compatible polymeric matrix.
  • the matrix may have a porous surface.
  • the implant material may be combined with a variety of matrix materials, including collagen.
  • the implant material may also contain biologically active substances, which may, for example, be bound to the matrix.
  • the implant material may be formed by known methods.
  • the invention also features methods for reforming and augmenting soft tissues.
  • the implant material may be implanted into soft tissue at a desired location. It may be accurately placed within soft tissue using a hand or orthoscopic device. In this manner, the implant material may be used to correct soft tissue defects, ( e . g . by plumping and expanding tissues).
  • the present invention pertains to a method for correcting contour defects within a human or animal which comprises the steps of applying a lattice sheet, e.g., a collagen-glycosaminoglycan matrix (CG matrix), within the surgical cavity. Blood vessels and mesenchymal cells are allowed to infiltrate the CG matrix from tissue within the cavity of a subject.
  • a lattice sheet e.g., a collagen-glycosaminoglycan matrix (CG matrix)
  • CG matrix collagen-glycosaminoglycan matrix
  • CG matrix synthetic membrane
  • the CG lattice serves as a supporting or scaffolding structure into which blood vessels and mesenchymal cells migrate from within a tissue cavity, a process referred to as "infiltration”. Infiltration is responsible for creating a new tissue, which replaces the CG matrix as it biodegrades.
  • glycosaminoglycans that may be suitable for use in this material include chondroitin 6-sulfate, chondroitin 4-sulfate, heparin, heparin sulfate, keratan sulfate, dermatan sulfate, chitin and chitosan.
  • FIG. 1 shows a schematic of the steps to the method of contour defect repair.
  • a surgical biopsy or lumpectomy procedure leaves a cavity in the tissue. Following the surgical biopsy or lumpectomy procedure, the wound cavity is inspected to ensure appropriate hemostasis.
  • the material is randomly packed into the cavity site. If FIG. 1C, the subcutaneous tissue and skin incisions are closed routinely.
  • FIG. 2 shows various forms of the lattice device of the present invention as a ball (2A), sheet (2B), coil (2C), and roll (2D).
  • the invention features devices and methods for making and using a permanent implant.
  • the present invention refers to a method of correcting contour defects on a human or animal. This invention overcomes many of the shortcomings presented by methods presently used to fill defects by using a preformed lattice structure for insertion into a surgical cavity.
  • the invention features devices and methods for soft tissue substitute.
  • the present invention features materials that may be implanted into soft body tissue for correction of soft tissue defects or for soft tissue augmentation.
  • the material comprises a biologically-compatible lattice or matrix.
  • the material may be combined with collagen, proteins or other matrix materials.
  • the matrix is absorbable, biocompatible, used as an acellular or cellular substrate, supports native tissue in-growth in three dimensions, and maintains the biomechanical properties of tissue (e.g. , breast tissue) during the postoperative period.
  • tissue e.g. , breast tissue
  • the highly porous nature of this product serves as a support structure into which vessels and mesenchymal cells from the wound site migrate. The cells and vessels create new tissue that replaces the collagen glycosaminoglycan as it biodegrades. Because cells invade the matrix material, fibrous encapsulation does not occur to any great degree.
  • a surgical biopsy or lumpectomy procedure is performed through an opening 50 in the tissue 70 of a patient that leaves a cavity 30 in the tissue 70.
  • the cavity 30 is inspected to ensure appropriate hemostasis.
  • the lattice device 10 is randomly packed into the cavity 30 through the opening 50.
  • the opening 50 in the subcutaneous tissue and skin 70 is closed using routine surgical procedures known in the art.
  • the device 10 comprising a lattice or matrix comprises a synthetic material having at least one layer.
  • at least one layer of the matrix comprises a protein.
  • the protein is selected from the group consisting of fibrin, collagen, glycosaminoglycan, and combinations thereof.
  • the device is a soft tissue implant material comprising biologically-compatible polymeric lattice having pores, said pores having dimensions effective to permit soft tissue to grow therein.
  • the pores comprise between about zero and about 90 percent of said implant material. More preferably, the pores comprise between about 20 and about 80 percent of said implant material. More preferably, the pores comprise between about 40 and about 60 percent of said implant material. Generally, the pores have a size of less than about 100 microns.
  • the implant material is constructed of collagen comprising between about 30% and about 65% of the implant material by volume.
  • the lattice has an overall thickness of from about 10 to about 200 mils, preferably 25 to about 100 mils.
  • the invention provides for a method of augmenting soft tissue comprising: (a) providing a biologically-compatible implant material comprised of biologically compatible polymeric lattice; and (b) implanting said implant material within a cavity 30 within soft tissue 70.
  • the lattice is in the form of a sheet.
  • the implanting step includes inserting the implant material subcutaneously into an area having a soft tissue contour defect in an amount sufficient to at least partially remove the contour defect.
  • the lattice is formed from a crosslinked collagen-glycosaminoglycan composite containing at least about 0.5% by weight glycosaminoglycan.
  • the collagen-glycosaminoglycan composite contains a sulfate-containing glycosaminoglycan.
  • the collagen-glycosaminoglycan composite contains from about 6% to about 12% by weight of said sulfate-containing glycosaminoglycan.
  • the sulfate-containing glycosaminoglycan is selected from chondroitin 6-sulfate, chondroitin 4-sulfate, heparin, heparan sulfate, keratan sulfate or dermatan sulfate.
  • the collagen-glycosaminoglycan composite is crosslinked to an Mc value of between about 800 and about 60,000.
  • the sulfate-containing glycosaminoglycan is chondroitin 6-sulfate.
  • the collagen glycosaminoglycan material is placed directly into the wound following surgery to fill the cavity and correct contour defects resulting from the removal of tissue.
  • the product may be used at the time of the initial surgery, or in subsequent surgery.
  • the material used in this invention is preferably in sheet form.
  • the sheet may be randomly folded, rolled, cinched, or configured otherwise to fill the defect.
  • Other forms of the collagen glycosaminoglycan include forms such as blocks ( e . g ., 2.5 cm x 2.5 cm x 1 cm blocks), spheres, and other configurations.
  • the sheet or other form may be cut to the size required to fill the defect.
  • the present invention is a soft tissue implant material comprising biologically-compatible polymeric matrix.
  • the matrix may have a porous surface.
  • the implant material may be combined with a variety of matrix materials, including collagen.
  • the implant material may also contain bioactive substances, which may, for example, be grafted to the matrix.
  • the implant material may be formed by known methods.
  • the bioactive compounds of the invention are administered in a therapeutically effective amount, i . e ., a dosage sufficient to effect treatment, which may vary depending on the individual and condition being treated.
  • a therapeutically effective daily dose may be from 0.1 to 100 mg/kg of body weight per day of drug.
  • Many conditions may respond to administration of a total dosage of between about 1 and about 30 mg/kg of body weight per day, or between about 70 mg and 2100 mg per day for a 70 kg person.
  • Other dosages may be administered without departure from the present invention.
  • the devices of the present invention may also be used for localized delivery of various drugs and other biologically active agents.
  • Biologically active agents such as growth factors may be delivered from the device to a local tissue site in order to facilitate tissue healing and regeneration.
  • biologically active agent or “active agent” as used herein refers to organic molecules that exert biological effects in vivo.
  • active agents include, without limitation, enzymes, receptor antagonists or agonists, hormones, growth factors, angiogenic factors, autogenous bone marrow, antibiotics, antimicrobial agents and antibodies.
  • active agent is also intended to encompass various cell types and genes that can be incorporated into the devices of the invention.
  • active agent is also intended to encompass combinations or mixtures of two or more active agents, as defined above.
  • TGFs transforming growth factors
  • FGFs fibroblast growth factors
  • PDGFs platelet derived growth factors
  • EGFs epidermal growth factors
  • CAPs connective tissue activated peptides
  • osteogenic factors and biologically active analogs, fragments, and derivatives of such growth factors.
  • TGF transforming growth factor
  • FGFs fibroblast growth factors
  • PDGFs platelet derived growth factors
  • EGFs epidermal growth factors
  • CAPs connective tissue activated peptides
  • osteogenic factors and biologically active analogs, fragments, and derivatives of such growth factors.
  • TGF transforming growth factor
  • TGF transforming growth factor
  • TGF supergene family include the beta transforming growth factors (for example, TGF- ⁇ 1, TGF- ⁇ 2, TGF- ⁇ 3); bone morphogenetic proteins (for example, BMP-1, BMP-2, BMP-3, BMP-4, BMP-5, BMP-6, BMP-7, BMP-8, BMP-9); heparin-binding growth factors (for example, fibroblast growth factor (FGF), epidermal growth factor (EGF), platelet-derived growth factor (PDGF), insulin-like growth factor (IGF)); inhibins (for example, Inhibin A, Inhibin B); growth differentiating factors (for example, GDF-1); and Activins (for example, Activin A, Activin B, Activin AB).
  • FGF fibroblast growth factor
  • EGF epidermal growth factor
  • PDGF platelet-derived growth factor
  • IGF insulin-like growth factor
  • FGF fibroblast growth factor
  • Inhibin A for example, Inhibin B
  • growth differentiating factors for
  • Steroidal anti-inflammatories can be used to decrease inflammation to the implanted device. These factors are known to those skilled in the art and are available commercially or described in the literature.
  • the active agents are incorporated to between 1 % and 30% by weight, although the factors can be incorporated to a weight percentage between 0.01 and 95 weight percentage. Active agents can be incorporated into the device and released over time by diffusion and/or degradation of the device or incorporated within the device, or some combination thereof. The type and amount of active agent used will depend, among other factors, on the particular site and condition to be treated and the biological activity and pharmacokinetics of the active agent selected.
  • Cytotoxic and immunosuppressive drugs may constitute an additional class of drugs for which the implant devices of the invention may be useful. These agents are commonly used to treat hyperproliferative diseases such as psoriasis, as well as for immune diseases such as bullous dermatoses and leukocytoclastic vasculitis. Examples of such compounds include, but are not limited to, antimetabolites such as methotrexate, azathioprine, fluorouracil, hydroxyurea, 6-thioquanine, mycophenolate, chlorambucil, vinicristine, vinblasrine and dactinomycin.
  • antimetabolites such as methotrexate, azathioprine, fluorouracil, hydroxyurea, 6-thioquanine, mycophenolate, chlorambucil, vinicristine, vinblasrine and dactinomycin.
  • alkylating agents such as cyclophosphamide, mechloroethamine hydrochloride, carmustine, taxol, tacrolimus and vinblastine are additional examples of useful biological agents, as are dapsone and sulfasalazine.
  • Ascomycins such as Cyclosporine, FK506 (tacrolimus), and rapamycin (e.g., U.S. Pat. No. 5,912,253) and analogs of such compounds are of particular interest ( e.g. , Mollinson et al ., Current Pharm. Design 4(5):367-380 (1998); U.S. Pat. Nos.
  • Cyclosporins include cyclosporin A, B, C, D, G and M. See, e . g ., U.S. Pat. Nos. 6,007,840; and 6,004,973.
  • Another aspect of the invention comprises delivery of taxane- and taxoid anticancer compositions, which are particularly useful for inhibiting growth of cancer cells.
  • the implant devices may include bioactive agents useful for treating conditions such as lupus erythematosus (both discoid and systemic), cutaneous dermatomyositis, porphyria cutanea tarda and polymorphous light eruption.
  • Agents useful for treating such conditions include, for example, quinine, chloroquine, hydroxychloroquine, and quinacrine.
  • the implant devices of the invention may also useful for transdermal delivery of antiinfective agents.
  • antibacterial, antifungal and antiviral agents may be used with the implant devices.
  • Antibacterial agents may be useful for treating conditions such as acne, cutaneous infections, and the like.
  • Antifungal agents may be used to treat tinea corporis, tinea pedis, onychomycosis, candidiasis, tinea versicolor, and the like.
  • antifungal agents include, but are not limited to, azole antifungals such as itraconazole, myconazole and fluconazole.
  • antiviral agents include, but are not limited to, acyclovir, famciclovir, and valacyclovir. Such agents may be useful for treating viral diseases, e.g., herpes.
  • bioactive agent for which the implant devices of the invention may include are the antihistamines. These agents are useful for treating conditions such as pruritus due to urticaria, atopic dermatitis, contact dermatitis, psoriasis, and many others. Examples of such reagents include, for example, terfenadine, astemizole, lorotadine, cetirizine, acrivastine, temelastine, cimetidine, ranitidine, famotidine, nizatidine, and the like. Tricyclic antidepressants may also be delivered.
  • Pain relief agents and local anesthetics constitute another class of compounds for which the implant devices of the invention may enhance treatment.
  • Lidocaine, bupibacaine, novocaine, procaine, tetracaine, benzocaine, cocaine, and the opiates are among the compounds that may be used with the implant devices of the invention.
  • the bioactive agent formulation comprises a cardiac drug including, but is not limited to: angiogenic factors, growth factors, calcium channel blockers, antihypertensive agents, inotropic agents, antiatherogenic agents, anti-coagulants, beta-blockers, anti-arrhythmic agents, anti-inflammatory agents, sympathomimetic agents, phosphodiesterase inhibitors, diuretics, vasodilators, thrombolytic agents, cardiac glycosides, antibiotics, antiviral agents, antifungal agents, agents that inhibit protozoans, antineoplastic agents, and steroids.
  • a cardiac drug including, but is not limited to: angiogenic factors, growth factors, calcium channel blockers, antihypertensive agents, inotropic agents, antiatherogenic agents, anti-coagulants, beta-blockers, anti-arrhythmic agents, anti-inflammatory agents, sympathomimetic agents, phosphodiesterase inhibitors, diuretics, vasodilators, thrombolytic agents, cardiac glycosides, antibiotics, antiviral agents,
  • anti-arrhythmia agent refers to any drug used to treat a disorder of rate, rhythm or conduction of electrical impulses within the heart.
  • angiogenic agent means any compdund that promotes growth of new blood vessels.
  • Angiogenic factors include, but are not limited to, a fibroblast growth factor, e.g., basic fibroblast growth factor (bFGF), and acidic fibroblast growth factor, e.g., FGF-1, FGF-2, FGF-3, FGF-4, recombinant human FGF (U.S. Pat. No.
  • VEGF vascular endothelial cell growth factor
  • VEGF-alpha transforming growth factor-alpha
  • platelet derived growth factor an endothelial mitogenic growth factor
  • platelet activating factor tumor necrosis factor-alpha
  • angiogenin a prostaglandin, including, but not limited to PGE1, PGE2; placental growth factor; GCSF (granulocyte colony stimulating factor); HGF (hepatocyte growth factor); IL-8; vascular permeability factor; epidermal growth factor; substance P; bradykinin; angiogenin; angiotensin II; proliferin; insulin like growth factor-1; nicotinamide; a stimulator of nitric oxide synthase; estrogen, including, but not limited to, estradiol (E2), estriol (E3),
  • Angiogenic factors further include functional analogs and derivatives of any of the aforementioned angiogenic factors.
  • Derivatives include polypeptide angiogenic factors having an amino acid sequence that differs from the native or wild-type amino acid sequence, including conservative amino acid differences (e.g., serine/threonine, asparagine/glutamine, alanine/valine, leucine/isoleucine, phenylalanine/tryptophan, lysine/arginine, aspartic acid/glutamic acid substitutions); truncations; insertions; deletions; and the like, that do not substantially adversely affect, and that may increase, the angiogenic property of the angiogenic factor.
  • conservative amino acid differences e.g., serine/threonine, asparagine/glutamine, alanine/valine, leucine/isoleucine, phenylalanine/tryptophan, lysine/arginine, aspartic acid/
  • Angiogenic factors include factors modified by polyethylene glycol modifications; acylation; acetylation; glycosylation; and the like.
  • An angiogenic factor may also be a polynucleotide that encodes the polypeptide angiogenic factor.
  • Such a polynucleotide may be a naked polynucleotide or may be incorporated into a vector, such as a viral vector system such as an adenovirus, adeno-associated virus or lentivirus systems.
  • Antibiotics are among the bioactive agents that may be useful when used with the implant devices of the invention, particularly those that act on invasive bacteria, such as Shigella, Salmonella, and Yersinia. Such compounds include, for example, norfloxacin, ciprofloxacin, trimethoprim, sulfamethyloxazole, and the like. Anti-neoplastic agents may also be delivered by the implant devices of the invention including, for example, cisplatin, methotrexate, taxol, fluorouracil, mercaptopurine, donorubicin, bleomycin, and the like.
  • Exemplary anti-inflammatory agents include, but are in no way limited to, corticoids such as cortisone and ACTH, dexamethasone, cortisol, interleukin-1 and its receptor antagonists, and antibodies to TGF-beta, to interleukin-1 (IL-1), and to interferon-gamma.
  • Exemplary anti-oxidants include, but are in no way limited to, vitamin C (ascorbic acid) and vitamin E.
  • Exemplary angiogenic factors include, but are in no way limited to, fibroblast growth factor and nerve growth factor.
  • Angiogenic growth factors which may be used in the device include, but are not limited to, Basic Fibroblast Growth Factor (bFGF), (also known as Heparin Binding Growth Factor-II and Fibroblast Growth Factor II), Acidic Fibroblast Growth Factor (aFGF), (also known as Heparin Binding Growth Factor-I and Fibroblast Growth Factor-I), Vascular Endothelial Growth Factor (VEGF), Platelet Derived Endothelial Cell Growth Factor BB (PDEGF-BB), Angiopoietin-1, Transforming Growth Factor Beta (TGF-Beta), Transforming Growth Factor Alpha (TGF-Alpha), Hepatocyte Growth Factor, Tumor Necrosis Factor-Alpha (TNF-Alpha), Angiogenin, Interleukin-8 (IL-8), Hypoxia Inducible Factor-I (H1F-1), Angiotensin-Converting Enzyme (ACE) Inhibitor Quinaprilat, Angiotropin,
  • the invention also features methods for reforming and augmenting soft tissues.
  • the implant material may be implanted into soft tissue at a desired location. In injectable form it may be accurately placed within soft tissue using a syringe or orthoscopic device. In this manner, the implant material may be used to correct soft tissue defects, ( e . g . by plumping and expanding tissues) remediate medical conditions such as incontinence, and for cosmetic procedures.
  • the present invention provides for methods for correcting contour defects within a human or animal which comprises the steps of applying a lattice sheet within the surgical cavity. Blood vessels and tissue cells are allowed to infiltrate the lattice from tissue within the cavity of a subject.
  • the present invention has many advantages.
  • the lattice is preferably completely synthetic and biodegradable over time. There is therefore no risk of disease transmission from donor to patient.
  • synthetic materials provide reproducible components and manufacturing methods.
  • the lattice is made up of multiple layers wherein the layers'are constructed from either proteinaceous or synthetic materials, or a combination wherein at least one layer is constructed from a proteinaceous material and at least one layer is constructed from a synthetic material.
  • the layers themselves can be constructed from all proteinaceous or all synthetic materials, or a combination of proteins and synthetic materials.
  • suitable proteins include alginates, fibrin, collagen, and glycosaminoglycan.
  • suitable synthetic materials include hydrogels, such as polyethylene glycol.
  • a preferred embodiment of the present invention employs a highly porous lattice comprised of collagen and glycosaminoglycan (referred to hereinafter as "GAG”), i . e . a collagen glycosaminoglycan matrix (referred to hereinafter as "CG matrix").
  • GAG glycosaminoglycan
  • CG matrix collagen glycosaminoglycan matrix
  • GAG Various forms of GAG which may be suitable for use in this material include chondroitin 6-sulfate, chondroitin 4-sulfate, heparin, heparin sulfate, keratan sulfate, dermatan sulfate, chitin and chitosan. See also US 5489304A1, US 5997895A1, WO9913902A1, and WO9706837A1.
  • the CG lattice serves as a supporting or scaffolding structure into which blood vessels and surrounding tissue cells migrate from within a tissue cavity, a process referred to as "infiltration". Infiltration is responsible for creating a new tissue, which replaces the lattice as it biodegrades.
  • lattice is used broadly herein to include any material that is in the form of a highly porous and permeable structure in which cells can migrate and proliferate.
  • Fibrous lattices should be construed broadly to include all lattices, which include material that is fibrous at the macroscopic, microscopic, or molecular level.
  • many polymeric foams comprise long organic molecules, which may have numerous side chains or extensive crosslinking.
  • the lattice of the present invention is not limited to collagen. Other fibrous proteins, other polymeric molecules, or sintered ceramics may also be suitable for prosthetic or other medical purposes.
  • the lattice serves as a temporary substitute for the tissue and can be any structure that has the following characteristics: the composition and structure of the lattice must be such that is does not provoke a substantial immune response from the graft recipient; the lattice must be sufficiently porous to permit blood vessels and cells from healthy tissue to migrate into the lattice; the lattice is biodegradable and the biodegradation must not proceed so rapidly that the lattice disappears before sufficient healing occurs.
  • CG matrix primarily crosslinking density, porosity and GAG content
  • specific conditions for forming a CG matrix suitable for use in the present invention are given below.
  • the skilled artisan will know of other conditions for forming CG matrices with variations of the above-mentioned parameters that are similarly suitable for use in the present invention.
  • certain applications of tissue regeneration may require matrices that degrade more slowly or more quickly.
  • the skilled artisan will be able to recognize applications where it is desirable to vary the properties of the CG matrix, and will be able to vary the parameters accordingly.
  • the present invention encompasses such variations in the CG matrix.
  • Collagen is a major protein constituent of connective tissues in vertebrate as well as invertebrate animals. It is often present in the form of macroscopic fibers that can be chemically and mechanically separated from non-collagenous tissue components.
  • Collagen derived from any source is suitable for use with this invention, including insoluble collagen, collagen soluble in acid, in neutral or basic aqueous solutions, as well as those collagens, which are commercially available.
  • Typical animal sources include calfskin, bovine Achilles tendon, cattle bones and rat tail tendon.
  • collagen Several levels of structural organization exist in collagen.
  • the primary structure consists of the complete sequence of amino acids.
  • Collagen is made up of 18 amino acids in relative amounts, which are well known for several animal species but in sequences and which are still not completely determined.
  • the total content of acidic, basic and hydroxylated amino acid residues far exceeds the content of lipophilic residues making collagen a hydrophilic protein. Because of this, polar solvents with high solubility parameters are good solvents for collagen.
  • mucopolysaccharide describes hexosamine-containing polysaccharides of animal origin. Another name often used for this class of compounds is glycosaminoglycans. Chemically, mucopolysaccharides are alternating copolymers made up of residues of hexosamine glycosidically bound and alternating in a more-or-less regular manner with either hexuronic acid or hexose moieties.
  • Typical sources of heparin include hog intestine, beef lung, bovine liver capsule and mouse skin.
  • Hyaluronic acid can be derived from rooster comb and human umbilical cord, whereas both chondroitin 4-sulfate and chondroitin 6-sulfate can be derived from bovine cartilage and shark cartilage.
  • Dermatan sulfate and heparan sulfate can be derived from hog mucosal tissues while keratan sulfate can be derived from the bovine cornea.
  • Suitable collagen can be derived from a number of animal sources, either in the form of a solution or in the form of a dispersion.
  • the invention relates to the use of composite materials formed by intimately contacting collagen with a mucopolysaccharide under conditions at which they form a reaction product and subsequently covalently crosslinking the reaction product.
  • Suitable mucopolysaccharides include, but are not limited to chondroitin 4-sulfate, chondroitin 6-sulfate, heparan sulfate, dermatan sulfate, keratan sulfate, heparin and hyaluronic acid.
  • Covalent crosslinking can be achieved by chemical, radiation, dehydrothermal or other covalent crosslinking techniques.
  • a suitable chemical technique is aldehyde crosslinking, but other chemical crosslinking reactants are equally suitable.
  • Dehydrothermal crosslinking which is preferred, is achieved by reducing the moisture level of the composites to a very low level, such as by subjecting the composite material to elevated temperatures and high vacuum.
  • Dehydrothermal crosslinking eliminates the necessity to add, and in the case of toxic materials such as aldehydes, to remove unreacted crosslinking agents; dehydrothermal crosslinking also produces composite materials containing a wider range of mucopolysaccharide content than is achieved with some chemical crosslinking techniques.
  • the products of such syntheses are collagen molecules or collagen fibrils with long mucopolysaccharide chains attached to them.
  • Such materials could include synthetic polymers such as the segmented polyurethanes, polyhydroxyethyl methacrylate and other "hydrogels", silicones, polyethylene terephthalate and polytetrafluoroethylene or modified natural polymers such as cellulose acetate or natural polymers such as elastin (the fibrous, insoluble, non-collagenous protein found in connective tissues such as the thoracic aorta and ligamentum nuchae) or pyrolytic carbon and other carbons which may have been treated thermally or by an electric arc.
  • synthetic polymers such as the segmented polyurethanes, polyhydroxyethyl methacrylate and other "hydrogels", silicones, polyethylene terephthalate and polytetrafluoroethylene or modified natural polymers such as cellulose acetate or natural polymers such as elastin (the fibrous, insoluble, non-collagenous protein found in connective tissues such as the thoracic aorta and ligamentum nuchae)
  • Such composites could be formed either by intimate mixing of the powdered solids or mixing of compatible solutions or dispersions of the two components or by coating with a mucopolysaccharide one of the materials mentioned in this paragraph. Irrespective of the method used to contact the mucopolysaccharide with the other material, the two components could be covalently bonded to form a material from which the mucopolysaccharide cannot be dissolved or extracted by contact with mucopolysaccharide solvents such as aqueous electrolytic solutions. Covalent bonding could be effected by a radiation grafting copolymerization technique using, for example, .gamma.-radiation from a cobalt-60 source.
  • blood-compatible materials could be prepared by bonding, using an adhesive, the crosslinked collagen-mucopolysaccharide composite in the form of a sheet, film, or other form onto a variety of substrates.
  • substrates would include synthetic polymers such as the segmented polyurethanes, polyhydroxyethyl methacrylate and other "hydrogels", silicones, polyethylene terephthalate and polytetrafluoroethylene or modified natural polymers such as cellulose acetate or natural polymers such as elastin or pyrolytic carbon and other carbons which may have been treated thermally or by an electric arc or metals such as vitalium, titanium and various steels.
  • a suitable adhesive would, for example, be a silicone rubber adhesive.
  • the method for producing the product of the present invention must make use of steps that are recognized as effective for inactivating viral and prion contamination. This gives the product a very high safety level while eliminating the inflammatory response. That is, the method for producing the product of the invention provides a product that is substantially free of viruses and prions without being physiologically incompatible.
  • the phrase "substantially free of viruses and prions" means that the product does not contain infection-effective amounts of viruses and prions.
  • the collagen devices of the present invention may be prepared by enzyme treatment, e.g., with ficin and/or pepsin for about 1 to 2 hours at a temperature of about 36.5°C to 37.5°C, an alkali treatment, e.g ., with an aqueous solution of 5% sodium hydroxide and 20% sodium sulfate at a pH of about 13 to 14, at a temperature of about 25°C to 30°C for a period of about 35 to 48 hours, or physiologically compatible collagen which is substantially free of active viruses and prions can be obtained from transgenic animals bred for the purpose of synthesizing human collagen in a readily harvestible form. See, e.g., U.S. Pat. No. 5,667,839.
  • the invention preferably comprises the use of collagen treated by a process sufficient to achieve at least a 4 log clearance of virus, more preferably at least a 6 log clearance of virus, and even more preferably at least an 8 log clearance of virus, as measured with a statistical confidence level of at least 95%. For example, if the concentration of virus before treatment is 10 7 and after treatment is 10 1 , then there has been an 6 log clearance of virus.
  • a collagen dispersion is first prepared in a manner well known in the art. One such preparation is taught in U.S. Pat. No. 3,157,524. Another suitable preparation of collagen is taught in U.S. Pat. No. 3,520,402. The product is preferably nonantigenic in addition to being noninfectious and physiologically compatible.
  • the matrix can include biocompatible and/or bioresorbable materials other than collagen, although collagen is most preferred.
  • Additional suitable polymers include, e . g ., biocompatible and/or bioresorbable lactides, glycolides, and copolymers thereof, polycaprolactones, polyethylene carbonate, tyrosine polycarbonates, tyrosine polyacids, and polyanhydrides.
  • the molecular weight of the polymer is preferably about 5000 to about 500,000.
  • polymer refers inter alia to polyalkyls, polyamino acids and polysaccharides. Additionally, for external or oral use, the polymer may be polyacrylic acid or carbopol.
  • synthetic polymer refers to polymers that are not naturally occurring and that are produced via chemical synthesis. As such, naturally occurring proteins such as collagen and naturally occurring polysaccharides such as hyaluronic acid are specifically excluded. Synthetic collagen, and synthetic hyaluronic acid, and their derivatives, are included. Synthetic polymers containing either nucleophilic or electrophilic groups are also referred to herein as "multifunctionally activated synthetic polymers".
  • multifunctionally activated refers to synthetic polymers which have, or have been chemically modified to have, two or more nucleophilic or electrophilic groups which are capable of reacting with one another (i . e ., the nucleophilic groups react with the electrophilic groups) to form covalent bonds.
  • Types of multifunctionally activated synthetic polymers include difunctionally activated, tetrafunctionally activated, and star-branched polymers.
  • glycosaminoglycans can additionally be incorporated into the compositions of the invention.
  • Glycosaminoglycans that can be derivatized according to either or both of the aforementioned methods include the following: hyaluronic acid, chondroitin sulfate A, chondroitin sulfate B (dermatan sulfate), chondroitin sulfate C, chitin (can be derivatized to chitosan), keratan sulfate, keratosulfate, and heparin.
  • collagen from any source may be used in the compositions of the invention; for example, collagen may be extracted and purified from human or other mammalian source, such as bovine or porcine corium and human placenta, or may be recombinantly or otherwise produced.
  • human or other mammalian source such as bovine or porcine corium and human placenta
  • the preparation of purified, substantially non-antigenic collagen in solution from bovine skin is well known in the art. See U.S. Pat. No. 5,428,022.
  • the term “collagen” or “collagen material” as used herein refers to all forms of collagen, including those that have been processed or otherwise modified.
  • Collagen of any type including, but not limited to, types I, II, III, IV, or any combination thereof, may be used in the compositions of the invention, although type I is generally preferred.
  • Either atelopeptide or telopeptide-containing collagen may be used; however, when collagen from a xenogeneic source, such as bovine collagen, is used, atelopeptide collagen is generally preferred, because of its reduced immunogenicity compared to telopeptide-containing collagen.
  • Collagen that has not been previously crosslinked by methods such as heat, irradiation, or chemical crosslinking agents is preferred for use in the compositions of the invention, although previously crosslinked collagen may be used.
  • Non-crosslinked atelopeptide fibrillar collagen is commercially available from Collagen Corporation (Palo Alto, Calif.) under the trademarks ZYDERM I COLLAGEN and ZYDERM II COLLAGEN, respectively.
  • Glutaraldehyde crosslinked atelopeptide fibrillar collagen is commercially available from Collagen Corporation under-the trademark ZYPLAST COLLAGEN.
  • the cavity is readied for application of the covering. Areas of tissue within the cavity that have been destroyed or damaged are surgically removed to prevent it from interfering with the healing process.
  • the CG matrix preferably in sheet form, is randomly stuffed into the cavity in a manner that minimizes the entrapment of air pockets between the tissue and the matrix.
  • the surgical opening is sutured or stapled closed using conventional techniques and then covered with a bandage.
  • Infiltration As defined herein, further refers to allowing a sufficient period of time for this migration of cells and blood vessels into the lattice.
  • the collagen lattice eventually is biodegraded by collagenase and other natural enzymes into non-toxic substances that are digested, utilized, or eliminated by normal bodily processes.
  • the lattice must retain its structural integrity until an adequate number of cells have reproduced within the lattice to regenerate the lost or removed tissue. If the lattice is biodegraded more quickly than this, it will be liquified and rendered useless before the cavity has healed.
  • the biodegradation rate should be roughly equal to approximately 25 to 30 days. This does not mean that the entire lattice should be biodegraded within 30 days. Instead, it indicates that a significant amount of biodegradation should commence within about 30 days, although remnants of the lattice may persist for several months or more. Routine experimentation by persons skilled in the art might indicate that this biodegradation rate should be modified somewhat for lattices that are seeded with cells, or for lattices that are used for purposes other than synthetic skin.
  • the biodegradation rate of a collagen lattice may be decreased (i.e., the lattice will endure for a longer period of time after grafting onto a wound) by increasing the collagen crosslinking density, by increasing the content of GAG that is crosslinked with collagen, or by decreasing the porosity of the lattice.
  • the lattice device should be sufficiently tough and strong to withstand suturing without tearing, and to prevent or limit tearing if subjected to accidental stresses caused by bandaging or medical operations or by patient movement.
  • the two most important indices of strength of a lattice are tensile strength (which measures how much force is required to pull apart a specimen with a known cross-sectional area) and fracture energy (which measures how much work is required to create a tear of a given size).
  • the strength of the lattice may be increased by increasing the crosslinking density or by decreasing the porosity of the lattice.
  • the synthetic collagen lattice should resemble the collagen matrix that exists naturally within the type of tissue that is to be regenerated. This spatial arrangement will promote the growth of cells in orderly patterns that resemble undamaged tissue, thereby reducing scarring and promoting proper functioning of the regenerated tissue.
  • CG matrices refer to CG matrices into which cells (preferably harvested from a wound free site on the patient's body) have been introduced. Each cell that survives the seeding process can reproduce and multiply, thereby hastening the formation of a cavity-filling tissue.
  • Preferred cells are adipocytes (fat cells) and/or preadipocytes (fat cell precursors that differentiate into adipocytes) derived from fat storage areas in the body or fat cells derived from bone marrow, stem cells, or mesenchymal cells that are harvested and developed under laboratory conditions and seeded onto the matrix prior to implantation.
  • CG matrices are described in U.S. Pat. No. 4,060,081, the teachings of which are incorporated herein by reference in its entirety. Matrices that have been seeded are referred to as “cellular” while unseeded matrices are referred to as “acellular”.
  • Seeded CG matrices may be autologous, i.e. matrices seeded with cells obtained from the human or animal having the bum or wound, or they may be heterologous, i.e. seeded with cells obtained from a donor.
  • cells being used to seed a CG matrix may undergo genetic manipulation in order to prevent rejection or to change the cell's phenotype in some beneficial manner. Genetic manipulation includes introducing genetic matter into the cells so that the protein gene product or products are expressed in sufficient quantities to cause the desired change in phenotype.
  • An example of suitable genetic matter includes the gene encoding for a growth factor along with the requisite control elements.
  • Implants of the invention may also include radio-opaque materials or radio-opaque elements, so that the biopsy site may be detected both with ultrasound and with X-ray or other radiographic imaging techniques.
  • Radiopaque materials and markers may include metal objects such as clips, bands, strips, coils, and other objects made from radiopaque metals and metal alloys, and may also include powders or particulate masses of radiopaque materials.
  • Radiopaque markers may be of any suitable shape or size, and are typically formed in a recognizable shape not naturally found within a patient's body, such as a star, square, rectangular, geometric, gamma, letter, coil or loop shape.
  • Suitable radiopaque materials include stainless steel, platinum, gold, iridium, tantalum, tungsten, silver, rhodium, nickel, bismuth, other radiopaque metals, alloys and oxides of these metals, barium salts, iodine salts, iodinated materials, and combinations of these.
  • the implant of the invention may also include MRI-detectable materials or markers, so that the biopsy site may be detected both with ultrasound and with MRI or other imaging techniques.
  • MRI contrast agents such as gadolinium and gadolinium compounds, for example, are suitable for use with ultrasound-detectable biopsy marker materials embodying features of the invention.
  • Colorants such as dyes (e.g., methylene blue and carbon black) and pigments (e.g., barium sulfate), may also be included in ultrasound-detectable biopsy marker materials embodying features of the invention.
  • the biocompatibility of two scaffolds is investigated in a porcine model.
  • the sample materials designated H16 and IM by Integra, (blocks measuring 2.5 x 2.5 x 1.0 cm 3 ) are placed in the mammary tissue of three pigs and evaluated at explant time periods of 7, 21, and 60 days.
  • histology reveals giant cell infiltration with minimal fibrous tissue invasion.
  • Neutrophil and giant cell infiltration is ongoing at 21 days with moderate to significant fibrous tissue invasion.
  • invasion of well-organized granulation tissue is observed.
  • the histopathology findings are considered positive and indicated a need for follow-up studies with an extended duration.
  • Implant site volume is maintained between days 14 and 42. Between days 42 and 179, the implants in this pilot study followed one of two paths depending on the animal in which they are implanted.
  • tissue reactions within the Integra matrices are associated with the normal absorptive process of the material and are no greater than slight to moderate.
  • Site volume maintenance correlated with matrix absorption as the material absorbed, the sites became smaller. Although the one site with total absorption at day 364 has a greater amount of fibrous connective tissue than the empty control sites at that period, the absolute amount of new tissue is considerably less than the volume of material originally introduced into the site.
  • the degree of calcification of unabsorbed matrix appears to increase with in vivo residence.
  • scaffold materials including the Integra product (identified as Avagen below), are evaluated for in vitro biocompatibility with human preadipocytes. All of these scaffolds are seeded with human subcutaneous preadipocytes at 5X10 3 cell/cm 2 2.5X10 4 cells/cm 2 and 2.5X10 5 cells/cm 2 . A fluorometric assay is used to measures the total DNA in the scaffolds which indirectly estimates the number of cells. Proliferation of preadipocytes is observed with all scaffolds and is dependent on the initial seeding density and type of scaffold.
  • a decrease in cell number and leptin secretion is observed in non-woven materials following adipocyte differentiation that may be due to the relatively rapid degradation rate of vicryl and the reduction in pH value which had an effect on pre-adipocyte biological activity.
  • Hematoxylin and eosin (H&E) staining reveals the penetration of preadipocytes through the scaffolds after a three-week incubation.
  • fluorescence labeling of adipocytes with Nile Red and histological staining with oil red O reveals that most of the mature adipocytes is on the surface of the scaffolds.
  • Pre-adipocytes proliferated and differentiate well on both CBC foams and Avagen.TM

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Dermatology (AREA)
  • Medicinal Chemistry (AREA)
  • Oral & Maxillofacial Surgery (AREA)
  • Transplantation (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Dispersion Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Composite Materials (AREA)
  • Materials Engineering (AREA)
  • Materials For Medical Uses (AREA)
EP04258168A 2003-12-30 2004-12-29 Matrice de collagène pour l'augmentation de tissus mous Withdrawn EP1555035A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US748894 2003-12-30
US10/748,894 US20050142161A1 (en) 2003-12-30 2003-12-30 Collagen matrix for soft tissue augmentation

Publications (1)

Publication Number Publication Date
EP1555035A2 true EP1555035A2 (fr) 2005-07-20

Family

ID=34620640

Family Applications (1)

Application Number Title Priority Date Filing Date
EP04258168A Withdrawn EP1555035A2 (fr) 2003-12-30 2004-12-29 Matrice de collagène pour l'augmentation de tissus mous

Country Status (5)

Country Link
US (1) US20050142161A1 (fr)
EP (1) EP1555035A2 (fr)
JP (1) JP2005193055A (fr)
AU (1) AU2004242529A1 (fr)
CA (1) CA2491788A1 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2067494A1 (fr) * 2007-12-04 2009-06-10 Charité-Universitätsmedizin Berlin Structure tubulaire ou en feuille formé d'un matériau biocompatible élastique et son utilisation
WO2009151414A1 (fr) * 2008-06-10 2009-12-17 Endomedix, Inc. Feuillet adhérent biocompatible pour suture des tissus
US7854923B2 (en) 2006-04-18 2010-12-21 Endomedix, Inc. Biopolymer system for tissue sealing
AU2009300354B2 (en) * 2008-09-23 2014-02-06 Senorx, Inc. Porous bioabsorbable implant
US10517988B1 (en) 2018-11-19 2019-12-31 Endomedix, Inc. Methods and compositions for achieving hemostasis and stable blood clot formation

Families Citing this family (76)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8668737B2 (en) 1997-10-10 2014-03-11 Senorx, Inc. Tissue marking implant
US9820824B2 (en) 1999-02-02 2017-11-21 Senorx, Inc. Deployment of polysaccharide markers for treating a site within a patent
US8498693B2 (en) 1999-02-02 2013-07-30 Senorx, Inc. Intracorporeal marker and marker delivery device
US8361082B2 (en) 1999-02-02 2013-01-29 Senorx, Inc. Marker delivery device with releasable plug
US20090216118A1 (en) 2007-07-26 2009-08-27 Senorx, Inc. Polysaccharide markers
CA2446993C (fr) 2000-11-20 2009-06-02 Senorx, Inc. Marqueurs de sites tissulaires pour imagerie in vivo
AU2003230321B8 (en) * 2002-05-09 2009-07-30 Medigenes A pharmaceutical composition for treatment of wounds containing blood plasma or serum
US20060036158A1 (en) 2003-11-17 2006-02-16 Inrad, Inc. Self-contained, self-piercing, side-expelling marking apparatus
US7877133B2 (en) 2003-05-23 2011-01-25 Senorx, Inc. Marker or filler forming fluid
US20050273002A1 (en) 2004-06-04 2005-12-08 Goosen Ryan L Multi-mode imaging marker
US8728463B2 (en) 2005-03-11 2014-05-20 Wake Forest University Health Science Production of tissue engineered digits and limbs
US20060204445A1 (en) * 2005-03-11 2006-09-14 Anthony Atala Cell scaffold matrices with image contrast agents
EP1863547B1 (fr) * 2005-03-11 2016-05-11 Wake Forest University Health Sciences Vaisseaux sanguins realises par genie tissulaire
AU2006223112B2 (en) 2005-03-11 2011-12-01 Wake Forest University Health Sciences Production of tissue engineered heart valves
US20060204539A1 (en) * 2005-03-11 2006-09-14 Anthony Atala Electrospun cell matrices
US7531503B2 (en) * 2005-03-11 2009-05-12 Wake Forest University Health Sciences Cell scaffold matrices with incorporated therapeutic agents
US10357328B2 (en) 2005-04-20 2019-07-23 Bard Peripheral Vascular, Inc. and Bard Shannon Limited Marking device with retractable cannula
US7323184B2 (en) * 2005-08-22 2008-01-29 Healagenics, Inc. Compositions and methods for the treatment of wounds and the reduction of scar formation
FR2894146B1 (fr) * 2005-12-07 2008-06-06 Ifremer Utilisation d'un polysaccharide excrete par l'espece vibrio diabolicus a des fins d'ingenierie des tissus conjonctifs non mineralises
WO2008073965A2 (fr) 2006-12-12 2008-06-19 C.R. Bard Inc. Marqueur tissulaire pour mode d'imagerie multiple
EP2101670B1 (fr) 2006-12-18 2013-07-31 C.R.Bard, Inc. Marqueur de biopsie présentant des propriétés d'imagerie générées in situ
US20080260794A1 (en) * 2007-02-12 2008-10-23 Lauritzen Nels J Collagen products and methods for producing collagen products
US9056151B2 (en) * 2007-02-12 2015-06-16 Warsaw Orthopedic, Inc. Methods for collagen processing and products using processed collagen
US8932619B2 (en) * 2007-06-27 2015-01-13 Sofradim Production Dural repair material
US20090068250A1 (en) * 2007-09-07 2009-03-12 Philippe Gravagna Bioresorbable and biocompatible compounds for surgical use
US9308068B2 (en) 2007-12-03 2016-04-12 Sofradim Production Implant for parastomal hernia
WO2009156866A2 (fr) 2008-06-27 2009-12-30 Sofradim Production Implant biosynthétique pour réparation de tissu mou
WO2010053918A1 (fr) * 2008-11-05 2010-05-14 Hancock Jaffe Laboratories, Inc. Composite contenant du collagène et de l'élastine en tant qu'expanseur dermique et agent de remplissage tissulaire
WO2010077244A1 (fr) 2008-12-30 2010-07-08 C.R. Bard Inc. Dispositif d'administration de marqueurs destiné à placer des marqueurs dans des tissus
JP2010227172A (ja) * 2009-03-26 2010-10-14 Terumo Corp 軟組織増大材料
FR2949688B1 (fr) 2009-09-04 2012-08-24 Sofradim Production Tissu avec picots revetu d'une couche microporeuse bioresorbable
US8790699B2 (en) 2010-04-23 2014-07-29 Warsaw Orthpedic, Inc. Foam-formed collagen strand
US8460691B2 (en) 2010-04-23 2013-06-11 Warsaw Orthopedic, Inc. Fenestrated wound repair scaffold
US20110295151A1 (en) 2010-05-26 2011-12-01 Bakos Gregory J Enteroendocrine Manipulation for Metabolic Effect
CN102120033A (zh) * 2011-03-07 2011-07-13 温州医学院 促口腔额面部多种创伤修复的复合生长因子的胶原蛋白缓释载体材料及其制备方法
FR2972626B1 (fr) 2011-03-16 2014-04-11 Sofradim Production Prothese comprenant un tricot tridimensionnel et ajoure
FR2977790B1 (fr) 2011-07-13 2013-07-19 Sofradim Production Prothese pour hernie ombilicale
FR2977789B1 (fr) 2011-07-13 2013-07-19 Sofradim Production Prothese pour hernie ombilicale
AU2012313984B2 (en) 2011-09-30 2016-02-11 Covidien Lp Reversible stiffening of light weight mesh
CN103841959A (zh) 2011-09-30 2014-06-04 索弗拉狄姆产品公司 用于输送治疗剂的多层植入物
FR2985170B1 (fr) 2011-12-29 2014-01-24 Sofradim Production Prothese pour hernie inguinale
FR2985271B1 (fr) 2011-12-29 2014-01-24 Sofradim Production Tricot a picots
EP3461508A1 (fr) 2012-01-24 2019-04-03 LifeCell Corporation Matrices tissulaires allongées
FR2994185B1 (fr) 2012-08-02 2015-07-31 Sofradim Production Procede de preparation d’une couche poreuse a base de chitosane
FR2995778B1 (fr) 2012-09-25 2015-06-26 Sofradim Production Prothese de renfort de la paroi abdominale et procede de fabrication
FR2995779B1 (fr) 2012-09-25 2015-09-25 Sofradim Production Prothese comprenant un treillis et un moyen de consolidation
FR2995788B1 (fr) 2012-09-25 2014-09-26 Sofradim Production Patch hemostatique et procede de preparation
WO2014049446A2 (fr) 2012-09-28 2014-04-03 Sofradim Production Emballage pour dispositif de réparation des hernies
US8940684B2 (en) 2012-11-19 2015-01-27 Mimedx Group, Inc. Cross-linked collagen comprising an antifungal agent
US9155799B2 (en) 2012-11-19 2015-10-13 Mimedx Group, Inc. Cross-linked collagen with at least one bound antimicrobial agent for in vivo release of the agent
US8946163B2 (en) * 2012-11-19 2015-02-03 Mimedx Group, Inc. Cross-linked collagen comprising metallic anticancer agents
US10029030B2 (en) 2013-03-15 2018-07-24 Mimedx Group, Inc. Molded placental tissue compositions and methods of making and using the same
US10335433B2 (en) 2013-04-10 2019-07-02 Mimedx Group, Inc. NDGA polymers and metal complexes thereof
US9446142B2 (en) 2013-05-28 2016-09-20 Mimedx Group, Inc. Polymer chelator conjugates
FR3006578B1 (fr) 2013-06-07 2015-05-29 Sofradim Production Prothese a base d’un textile pour voie laparoscopique
FR3006581B1 (fr) 2013-06-07 2016-07-22 Sofradim Production Prothese a base d’un textile pour voie laparoscopique
US10449220B2 (en) 2013-08-30 2019-10-22 Mimedx Group, Inc. Micronized placental compositions comprising a chelator
USD716451S1 (en) 2013-09-24 2014-10-28 C. R. Bard, Inc. Tissue marker for intracorporeal site identification
USD715942S1 (en) 2013-09-24 2014-10-21 C. R. Bard, Inc. Tissue marker for intracorporeal site identification
USD716450S1 (en) 2013-09-24 2014-10-28 C. R. Bard, Inc. Tissue marker for intracorporeal site identification
USD715442S1 (en) 2013-09-24 2014-10-14 C. R. Bard, Inc. Tissue marker for intracorporeal site identification
US10092679B2 (en) 2013-10-18 2018-10-09 Wake Forest University Health Sciences Laminous vascular constructs combining cell sheet engineering and electrospinning technologies
EP3000489B1 (fr) 2014-09-24 2017-04-05 Sofradim Production Procédé de préparation d'un film barrière anti-adhésion
EP3000433B1 (fr) 2014-09-29 2022-09-21 Sofradim Production Dispositif pour introduire une prothèse pour le traitement de la hernie dans une incision et prothèse textile flexible
EP3000432B1 (fr) 2014-09-29 2022-05-04 Sofradim Production Prothèse à base textile pour le traitement d'une hernie inguinale
EP3029189B1 (fr) 2014-12-05 2021-08-11 Sofradim Production Tricot poreux prothétique, procédé pour son obtention et prothèse à hernie
EP3059255B1 (fr) 2015-02-17 2020-05-13 Sofradim Production Méthode pour préparer une matrice de chitosane comprenant un élément de renfort fibreux
EP3085337B1 (fr) 2015-04-24 2022-09-14 Sofradim Production Prothèse pour supporter une structure mammaire
EP3106185B1 (fr) 2015-06-19 2018-04-25 Sofradim Production Prothèse synthétique comprenant un tricot et un film non poreux et méthode pour la former
EP3195830B1 (fr) 2016-01-25 2020-11-18 Sofradim Production Prothèse de réparation de hernie
EP3312325B1 (fr) 2016-10-21 2021-09-22 Sofradim Production Méthode pour la fabrication un treillis avec suture crantée attachée et treillis ainsi obtenu
EP3398554A1 (fr) 2017-05-02 2018-11-07 Sofradim Production Prothèse pour réparation de hernie inguinale
EP3618882A4 (fr) 2017-05-05 2021-01-06 Ottawa Heart Institute Research Corporation Compositions d'hydrogel biocompatible et utilisations correspondantes
EP3653171A1 (fr) 2018-11-16 2020-05-20 Sofradim Production Implants conçus pour la réparation de tissus mous
US11090412B2 (en) * 2018-12-21 2021-08-17 Zavation Medical Products Llc Bone repair composition and kit
WO2020255113A1 (fr) * 2019-06-20 2020-12-24 Datum Biotech Ltd. Structure implantable ayant une membrane de collagène

Family Cites Families (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3157524A (en) * 1960-10-25 1964-11-17 Ethicon Inc Preparation of collagen sponge
US3520402A (en) * 1967-08-30 1970-07-14 Ethicon Inc Purified collagen fibrils
US4280954A (en) * 1975-07-15 1981-07-28 Massachusetts Institute Of Technology Crosslinked collagen-mucopolysaccharide composite materials
US4060081A (en) * 1975-07-15 1977-11-29 Massachusetts Institute Of Technology Multilayer membrane useful as synthetic skin
US4505266A (en) * 1981-10-26 1985-03-19 Massachusetts Institute Of Technology Method of using a fibrous lattice
US5604293A (en) * 1985-09-12 1997-02-18 Scios Inc. Recombinant human basic fibroblast growth factor
US4880429A (en) * 1987-07-20 1989-11-14 Stone Kevin R Prosthetic meniscus
US6007840A (en) * 1988-09-16 1999-12-28 Novartis Ag Pharmaceutical compositions comprising cyclosporins
US5643464A (en) * 1988-11-21 1997-07-01 Collagen Corporation Process for preparing a sterile, dry crosslinking agent
US5475052A (en) * 1988-11-21 1995-12-12 Collagen Corporation Collagen-synthetic polymer matrices prepared using a multiple step reaction
US5550187A (en) * 1988-11-21 1996-08-27 Collagen Corporation Method of preparing crosslinked biomaterial compositions for use in tissue augmentation
US5162430A (en) * 1988-11-21 1992-11-10 Collagen Corporation Collagen-polymer conjugates
US5510418A (en) * 1988-11-21 1996-04-23 Collagen Corporation Glycosaminoglycan-synthetic polymer conjugates
US5800541A (en) * 1988-11-21 1998-09-01 Collagen Corporation Collagen-synthetic polymer matrices prepared using a multiple step reaction
US5604294A (en) * 1991-09-05 1997-02-18 Luly; Jay R. Macrocyclic immunomodulators
IT1254704B (it) * 1991-12-18 1995-10-09 Mini Ricerca Scient Tecnolog Tessuto non tessuto essenzialmente costituito da derivati dell'acido ialuronico
US5428022A (en) * 1992-07-29 1995-06-27 Collagen Corporation Composition of low type III content human placental collagen
US5667839A (en) * 1993-01-28 1997-09-16 Collagen Corporation Human recombinant collagen in the milk of transgenic animals
EP0690713A4 (fr) * 1993-03-17 1996-04-03 Abbott Lab Immunomodulateurs macrocycliques substitues contenant des amines alicycliques
EP0711298A1 (fr) * 1993-07-30 1996-05-15 Abbott Laboratories Macrolactames activees ayant des pouvoirs immunomodulateurs
US5612350A (en) * 1993-11-30 1997-03-18 Abbott Laboratories Macrocyclic immunomodulators with novel cyclohexyl ring replacements
SK78196A3 (en) * 1993-12-17 1997-02-05 Sandoz Ag Rapamycin demethoxy-derivatives, preparation method thereof and pharmaceutical agent containing them
CA2140053C (fr) * 1994-02-09 2000-04-04 Joel S. Rosenblatt Systeme a base de collagene pour l'administration de drogue injectable, et utilisation
US5489304A (en) * 1994-04-19 1996-02-06 Brigham & Women's Hospital Method of skin regeneration using a collagen-glycosaminoglycan matrix and cultured epithelial autograft
EP0713707A1 (fr) * 1994-11-23 1996-05-29 Collagen Corporation Composition de collagène insectable, réticulable in situ pour augmenter des tissus
AR004480A1 (es) * 1995-04-06 1998-12-16 Amico Derin C D Compuestos de ascomicina que poseen actividad antiinflamatoria, pro cedimiento para prepararlos, uso de dichos compuestos para preparar agentesfarmaceuticos y composiciones farmaceuticas que los incluyen
US5859031A (en) * 1995-06-07 1999-01-12 Gpi Nil Holdings, Inc. Small molecule inhibitors of rotamase enzyme activity
BE1009856A5 (fr) * 1995-07-14 1997-10-07 Sandoz Sa Composition pharmaceutique sous la forme d'une dispersion solide comprenant un macrolide et un vehicule.
US5752974A (en) * 1995-12-18 1998-05-19 Collagen Corporation Injectable or implantable biomaterials for filling or blocking lumens and voids of the body
CA2239775C (fr) * 1995-12-18 2008-07-15 Collagen Corporation Composition a base de polymeres reticules et leur procede d'utilisation
ATE434034T1 (de) * 1996-08-23 2009-07-15 Vegenics Ltd Rekombinanter vasculärer endothelzellen wachstumsfaktor d (vegf-d)
US5786378A (en) * 1996-09-25 1998-07-28 Gpi Nil Holdings, Inc. Heterocyclic thioesters
FR2757521B1 (fr) * 1996-12-24 1999-01-29 Rhone Poulenc Rorer Sa Nouveaux derives de cyclosporine, leur preparation et les compositions pharmaceutiques qui les contiennent
US5997895A (en) * 1997-09-16 1999-12-07 Integra Lifesciences Corporation Dural/meningeal repair product using collagen matrix
US20030199887A1 (en) * 2002-04-23 2003-10-23 David Ferrera Filamentous embolization device and method of use
US20040121943A1 (en) * 2002-12-20 2004-06-24 Wei-Cherng Hsu Drug-free biodegradable 3D porous collagen-glycosaminoglycan scaffold

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7854923B2 (en) 2006-04-18 2010-12-21 Endomedix, Inc. Biopolymer system for tissue sealing
US8513217B2 (en) 2006-04-18 2013-08-20 Endomedix, Inc. Biopolymer system for tissue sealing
US9259434B2 (en) 2006-04-18 2016-02-16 Endomedix, Inc. Biopolymer system for tissue sealing
US9731044B2 (en) 2006-04-18 2017-08-15 Endomedix, Inc. Biopolymer system for tissue sealing
EP2067494A1 (fr) * 2007-12-04 2009-06-10 Charité-Universitätsmedizin Berlin Structure tubulaire ou en feuille formé d'un matériau biocompatible élastique et son utilisation
WO2009151414A1 (fr) * 2008-06-10 2009-12-17 Endomedix, Inc. Feuillet adhérent biocompatible pour suture des tissus
AU2009300354B2 (en) * 2008-09-23 2014-02-06 Senorx, Inc. Porous bioabsorbable implant
US10517988B1 (en) 2018-11-19 2019-12-31 Endomedix, Inc. Methods and compositions for achieving hemostasis and stable blood clot formation
US11033654B2 (en) 2018-11-19 2021-06-15 Endomedix, Inc. Methods and compositions for achieving hemostasis and stable blood clot formation

Also Published As

Publication number Publication date
AU2004242529A1 (en) 2005-07-14
CA2491788A1 (fr) 2005-06-30
US20050142161A1 (en) 2005-06-30
JP2005193055A (ja) 2005-07-21

Similar Documents

Publication Publication Date Title
EP1555035A2 (fr) Matrice de collagène pour l'augmentation de tissus mous
Zantop et al. Extracellular matrix scaffolds are repopulated by bone marrow‐derived cells in a mouse model of achilles tendon reconstruction
Konofaos et al. Nerve repair by means of tubulization: past, present, future
Panduranga Rao Recent developments of collagen-based materials for medical applications and drug delivery systems
US10092676B2 (en) Biohybrid composite scaffold
US7919112B2 (en) Implantable tissue compositions and method
US6998418B1 (en) Acellular biological material chemically treated with genipin
US8535719B2 (en) Biohybrid elastomeric scaffolds and methods of use thereof
Lin et al. Injectable systems and implantable conduits for peripheral nerve repair
WO2006033686A1 (fr) Facteur angiogénique et matériau biocompatible
WO2004108179A1 (fr) Compositions destinees a la reparation et a la regeneration du tissu-mere humain
WO2001012240A1 (fr) Materiaux biologiques
WO2000016822A1 (fr) Reparations tissulaires et compositions a cet effet
US20190151510A1 (en) Trizonal membranes for periosteum regeneration
EP2344208B1 (fr) Conduit de croissance des nerfs périphériques
US20180200405A1 (en) Methods of preparing ecm scaffolds and hydrogels from colon
Yang et al. Characteristics and biocompatibility of a biodegradable genipin‐cross‐linked gelatin/β‐tricalcium phosphate reinforced nerve guide conduit
Liu et al. A novel use of genipin-fixed gelatin as extracellular matrix for peripheral nerve regeneration
US20090136553A1 (en) Triggerably dissolvable hollow fibers for controlled delivery
Luis et al. Evaluation of two biodegradable nerve guides for the reconstruction of the rat sciatic nerve
Kumar et al. Reconstruction of large ventrolateral hernia in a buffalo with acellular dermal matrix: a method for treating large hernias in animals-a case report
Al-Ebadi et al. Effect of acellular bovine pericardium and urinary bladder submucosa matrixes in reconstruction of ventro-lateral hernias in bucks; molecular evaluation
RU2808880C1 (ru) Биорезорбируемый имплантат кровеносных сосудов на основе нановолокон
Rizvi et al. Peripheral nerve regeneration in the presence of collagen fibers: Effect of removal of the distal nerve stump
Chen et al. Biodegradable medical implants

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR LV MK YU

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20070703